Ligand-activated peroxisome proliferator-activated receptor-δ and -γ inhibit lipopolysaccharide-primed release of high mobility group box 1 through upregulation of SIRT1

J S Hwang, W J Lee, E S Kang, S A Ham, T Yoo, K S Paek, D S Lim, J T Do, H G Seo, J S Hwang, W J Lee, E S Kang, S A Ham, T Yoo, K S Paek, D S Lim, J T Do, H G Seo

Abstract

Peroxisome proliferator-activated receptors (PPARs) inhibit lipopolysaccharide (LPS)-primed release of high mobility group box 1 (HMGB1), a late proinflammatory mediator, but the underlying molecular mechanism is not completely understood. In this study, we demonstrated that the inhibition of HMGB1 release by PPAR-δ and -γ is associated with the deacetylase activity of SIRT1. Ligand-activated PPAR-δ and -γ inhibited LPS-primed release of HMGB1, concomitant with elevation in SIRT1 expression and promoter activity. These effects were significantly reduced in the presence of small interfering (si)RNAs against PPAR, indicating that PPAR-δ and -γ are involved in both HMGB1 release and SIRT1 expression. In addition, modulation of SIRT1 expression and activity by siRNA or chemicals correspondingly influenced the effects of PPARs on HMGB1 release, suggesting a mechanism in which SIRT1 modulates HMGB1 release. Furthermore, we showed for the first time that HMGB1 acetylated in response to LPS or p300/CBP-associated factor (PCAF) is an effective substrate for SIRT1, and that deacetylation of HMGB1 is responsible for blockade of HMGB1 release in macrophages. Finally, acetylation of HMGB1 was elevated in mouse embryonic fibroblasts from SIRT1-knockout mice, whereas this increase was completely reversed by ectopic expression of SIRT1. These results indicate that PPAR-mediated upregulation of SIRT1 modulates the status of HMGB1 acetylation, which, in turn, has a critical role in the cellular response to inflammation through deacetylation-mediated regulation of HMGB1 release.

Figures

Figure 1
Figure 1
Ligand-activated PPARs inhibit LPS-induced release of HMGB1. (a) RAW 264.7 cells grown to 60% confluency were incubated in serum-free medium for 24 h, and then stimulated with LPS in the presence or absence of PPAR ligands for 24 h. (b and c) Cells were transfected with siRNA against PPAR-δ (b) or PPAR-γ (c), and grown for 38 h. After incubation in serum-free medium for 24 h, the cells were stimulated with LPS for 24 h in the presence or absence of GW501516 (b) or rosiglitazone (c). Equal volumes of conditioned media or aliquots of cell lysates were subjected to western blotting for determination of HMGB1 levels. An image analyzer was used to quantitate band intensity, and the ratios of HMGB1 to Ponceau S are shown. The results are expressed as means±S.E.M. (n=3). **P<0.01 compared with the untreated group; ##P<0.01 compared with the LPS-treated group; ††P<0.01 compared with the group treated with LPS+GW501516 or rosiglitazone. Ponceau S staining or β-actin was used as a loading control
Figure 2
Figure 2
Acetylation is involved in the regulation of HMGB1 release. (a) RAW 264.7 cells incubated in serum-free medium for 24 h were pretreated with resveratrol or sirtinol for 1 h, and then stimulated with LPS for 6 h (for detection of acetyl-HMGB1) or 24 h (for detection of HMGB1 released). (b) Cells transfected with SIRT1 siRNA or control siRNA were grown for 38 h, after which they were incubated in serum-free medium for 24 h, and then stimulated with LPS for 6 h (for detection of acetyl-HMGB1) or 24 h (for detection of released HMGB1). Cell lysates were pulled down with anti-HMGB1 and immunoblotted with anti-acetyl-lysine to detect acetylated HMGB1. Each membrane was then stripped and re-probed for total HMGB1, as a loading control. For determination of released HMGB1, equal volumes of conditioned media were subjected to western blot analysis; Ponceau S staining was used as a loading control. Whole-cell lysates were subjected to Western blot analysis to determine the expression levels of HMGB1 (a) and SIRT1 (b). An image analyzer was used to quantitate band intensity, and the fold changes in the acetyl-HMGB1 to HMGB1 or HMGB1 to Ponceau S ratio are shown. The results are expressed as means±S.E.M. (n=3). **P<0.01, *P<0.05 compared with the untreated group; ##P<0.01, #P<0.05 compared with the LPS-treated group. Ponceau S staining or β-actin was used as a loading control
Figure 3
Figure 3
SIRT1 mediates the effects of PPAR-δ and -γ in the inhibition of LPS-primed release of HMGB1 through deacetylation. (a) RAW 264.7 cells grown to 60% confluency were incubated with serum-free medium for 24 h and then stimulated with LPS for the indicated times. (b) Cells incubated for 24 h in serum-free medium were stimulated with LPS for 6 h in the presence or absence of the ligand indicated. (c and d) Cells were transfected with SIRT1 siRNA and grown for 38 h, after which they were stimulated with LPS for 6 h in the presence or absence of GW501516 (c) or rosiglitazone (d). Whole-cell lysates were immunoprecipitated with anti-HMGB1, and then acetylated HMGB1 was detected by western blotting with an anti–acetyl-lysine antibody. An image analyzer was used to quantitate band intensity, and the ratios of acetylated HMGB1 to total HMGB1 are shown. The results are expressed as means±S.E.M. (n=3). *P<0.05 compared with the untreated group; #P<0.05 compared with the LPS-treated group; †P<0.05 compared with the group treated with LPS+GW501516 or rosiglitazone
Figure 4
Figure 4
Ligand-activated PPAR-δ and -γ upregulate SIRT1 expression. (a) RAW 264.7 cells were treated with GW501516 or rosiglitazone for the indicated times. Total RNA was extracted, and SIRT1 mRNA levels were determined by real-time PCR. (b) Cells were treated with GW501516 or rosiglitazone for the indicated times. (c and d) Cells transfected with siRNA against PPAR-δ (c) or PPAR-γ (d) were treated with the indicated PPAR ligand for 3 h. Aliquots of protein from cell lysates were analyzed by western blotting with anti-SIRT1 or anti-β-actin antibody. (e) Cells pretreated with cycloheximide or actinomycin D for 30 min were incubated with 100 nM GW501516 or 10 μM rosiglitazone for 1 h. Total RNA was extracted and reverse transcribed into cDNA. Equal amounts of cDNA were diluted and amplified by real-time PCR. The fold change in SIRT1 cDNA relative to the GAPDH control was determined and plotted. (f) Cells transfected with a SIRT1 promoter-reporter construct and pSV β-gal were grown for 38 h and then exposed to the indicated ligand for 1 h. The luciferase activity was normalized to β-galactosidase activity, and the data are expressed as means±S.E.M. (n=5). *P<0.05 compared with the untreated group; #P<0.05 compared with the GW501516- or rosiglitazone-treated group
Figure 5
Figure 5
SIRT1 is essential for the inhibition of LPS-induced HMGB1 release by PPAR-δ and -γ. (a and b) RAW 264.7 cells pretreated with resveratrol (a) or sirtinol (b) for 1 h were stimulated with LPS in the presence or absence of GW501516. (c and d) Cells pretreated with resveratrol (c) or sirtinol (d) for 1 h were incubated with LPS in the presence or absence of rosiglitazone. After incubation for 24 h, equal volumes of conditioned media were analyzed by western blotting with an anti-HMGB1 antibody. An image analyzer was used to quantitate band intensity, and the ratios of HMGB1 to Ponceau S are shown. The results are expressed as means±S.E.M. (n=3). **P<0.01 compared with the untreated group; ##P<0.01 compared with the LPS-treated group; ††P<0.01 compared with the group treated with LPS+GW501516 or rosiglitazone. Ponceau S staining was used as a loading control
Figure 6
Figure 6
Downregulation of SIRT1 abrogates the prevention of PPAR-δ and -γ against HMGB1 release. (a) RAW 264.7 cells grown to 60% confluency were incubated with serum-free medium for 24 h, and then stimulated with LPS in the presence or absence of the indicated ligand for 24 h. Aliquots of protein from cell lysates were analyzed by western blotting with anti-SIRT1 or anti-β-actin antibody. (b and c) Cells transfected with siRNA against SIRT1 were stimulated with LPS in the presence or absence of the GW501516 (b) or rosiglitazone (c) for 24 h. Equal volumes of conditioned media were analyzed by western blotting with anti-HMGB1 antibody. An image analyzer was used to quantitate band intensity, and the fold changes in the HMGB1 to Ponceau S or SIRT1 to β-actin ratio are shown. The results are expressed as means±S.E.M. (n=3). **P<0.01 compared with the untreated group; ##P<0.01, #P<0.05 compared with the LPS-treated group; ††P<0.01 compared with the group treated with LPS+GW501516 or rosiglitazone. Ponceau S staining or β-actin was used as a loading control
Figure 7
Figure 7
SIRT1-mediated deacetylation of HMGB1 is a critical factor in the PPAR-δ/γ–mediated inhibition of HMGB1 release. (a) RAW 264.7 cells were transfected with empty vector (pcDNA3.1/Myc) or pcDNA3.1-Myc-SIRT1. (b) Cells were transfected with empty vector (pcDNA3.1/HA) or pcDNA3.1-HA-PCAF. (c) Cells were transfected with empty vector (pcDNA3.1), pcDNA3.1-Myc-SIRT1, or pcDNA3.1-HA-PCAF. After incubation for 38 h, cells were maintained in serum-free medium for 24 h, and then stimulated with or without LPS for 6 h (for detection of acetyl-HMGB1) or 24 h (for detection of released HMGB1). Cell lysates were pulled down with anti-HMGB1 and immunoblotted with anti-acetyl-lysine to detect acetylated HMGB1. Each membrane was then stripped and re-probed for HMGB1, as a loading control. For determination of released HMGB1, equal volumes of conditioned media were subjected to Western blot analysis; Ponceau S staining was used as a loading control. Whole-cell lysates were subjected to western blot analysis with an anti-HMGB1, anti-SIRT1, anti-Myc, or anti-HA antibody, as appropriate, to determine the expression levels of HMGB1 and SIRT1 (a and c) or PCAF (b and c)
Figure 8
Figure 8
Acetylated HMGB1 is an effective substrate for SIRT1. (a) HEK293T cells were transfected with empty vector (pcDNA3.1), pcDNA3.1-Flag-HMGB1, or pcDNA3.1-Myc-SIRT1. (b) HEK293T cells were transfected with pcDNA3.1-HA-PCAF or increasing amounts (2 μg and 4 μg) of pcDNA3.1-Myc-SIRT1. After incubation for 48 h, the cells were stimulated with (a) or without (b) LPS for 3 h. (c) Whole-cell lysates from SIRT1-knockout (SIRT1−/−) or wild-type (SIRT1+/+) MEFs were pulled down with anti-HMGB1 and analyzed by western blotting with anti-acetyl-lysine, anti-HMGB1, or anti-SIRT1 antibody to detect acetylated HMGB1 or total HMGB1 and SIRT1. (d) SIRT1−/− MEFs were transfected with empty vector (pcDNA3.1-Myc) or pcDNA3.1-Myc-SIRT1 and incubated for 48 h, after which the cells were harvested and subjected to immunoprecipitation with anti-HMGB1 antibody. Acetylated HMGB1 or total HMGB1 and SIRT1 were detected by western blot analysis. β-actin was used as a loading control

References

    1. 1Kliewer SA, Umesono K, Noonan DJ, Heyman RA, Evans RM. Convergence of 9-cis retinoic acid and peroxisome proliferator signalling pathways through heterodimer formation of their receptors. Nature 1992; 358: 771–774.
    1. 2Issemann I, Green S. Activation of a member of the steroid hormone receptor superfamily by peroxisome proliferators. Nature 1990; 347: 645–650.
    1. 3Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schütz G, Umesono K et al. The nuclear receptor superfamily: the second decade. Cell 1995; 83: 835–839.
    1. 4Tugwood JD, Issemann I, Anderson RG, Bundell KR, McPheat WL, Green S. The mouse peroxisome proliferator activated receptor recognizes a response element in the 5' flanking sequence of the rat acyl CoA oxidase gene. EMBO J 1992; 11: 433–439.
    1. 5Kidani Y, Bensinger SJ. Liver X receptor and peroxisome proliferator-activated receptor as integrators of lipid homeostasis and immunity. Immunol Rev 2012; 249: 72–83.
    1. 6Ramanan S, Kooshki M, Zhao W, Hsu FC, Robbins ME. PPARalpha ligands inhibit radiation-induced microglial inflammatory responses by negatively regulating NF-kappaB and AP-1 pathways. Free Radic Biol Med 2008; 45: 1695–1704.
    1. 7Kim HJ, Ham SA, Kim SU, Hwang JY, Kim JH, Chang KC et al. Transforming growth factor-beta1 is a molecular target for the peroxisome proliferator-activated receptor delta. Circ Res 2008; 102: 193–200.
    1. 8Chawla A, Barak Y, Nagy L, Liao D, Tontonoz P, Evans RM. PPAR-gamma dependent and independent effects on macrophage-gene expression in lipid metabolism and inflammation. Nat Med 2001; 7: 48–52.
    1. 9Hwang JS, Kang ES, Ham SA, Yoo T, Lee H, Paek KS et al. Activation of peroxisome proliferator-activated receptor γ by rosiglitazone inhibits lipopolysaccharide-induced release of high mobility group box 1. Mediators Inflamm 2012; 2012: 352807.
    1. 10Bustin M, Hopkins RB, Isenberg I. Immunological relatedness of high mobility group chromosomal proteins from calf thymus. J Biol Chem 1978; 253: 1694–1699.
    1. 11Pallier C, Scaffidi P, Chopineau-Proust S, Agresti A, Nordmann P, Bianchi ME et al. Association of chromatin proteins high mobility group box (HMGB) 1 and HMGB2 with mitotic chromosomes. Mol Biol Cell 2003; 14: 3414–3426.
    1. 12Stros M, Ozaki T, Bacikova A, Kageyama H, Nakagawara A. HMGB1 and HMGB2 cell-specifically down-regulate the p53- and p73-dependent sequence-specific transactivation from the human Bax gene promoter. J Biol Chem 2002; 277: 7157–7164.
    1. 13Lotze MT, Tracey KJ. High-mobility group box 1 protein (HMGB1): nuclear weapon in the immune arsenal. Nat Rev Immunol 2005; 5: 331–342.
    1. 14Müller S, Scaffidi P, Degryse B, Bonaldi T, Ronfani L, Agresti A et al. New EMBO members' review: the double life of HMGB1 chromatin protein: architectural factor and extracellular signal. EMBO J 2001; 20: 4337–4340.
    1. 15Bonaldi T, Talamo F, Scaffidi P, Ferrera D, Porto A, Bachi A et al. Monocytic cells hyperacetylate chromatin protein HMGB1 to redirect it towards secretion. EMBO J 2003; 22: 5551–5560.
    1. 16Youn JH, Shin JS. Nucleocytoplasmic shuttling of HMGB1 is regulated by phosphorylation that redirects it toward secretion. J Immunol 2006; 177: 7889–7897.
    1. 17Ito I, Fukazawa J, Yoshida M. Post-translational methylation of high mobility group box 1 (HMGB1) causes its cytoplasmic localization in neutrophils. J Biol Chem 2007; 282: 16336–16344.
    1. 18Gardella S, Andrei C, Ferrera D, Lotti LV, Torrisi MR, Bianchi ME et al. The nuclear protein HMGB1 is secreted by monocytes via a non-classical, vesicle-mediated secretory pathway. EMBO Rep 2002; 3: 995–1001.
    1. 19Andersson U, Tracey KJ. HMGB1 is a therapeutic target for sterile inflammation and infection. Annu Rev Immunol 2011; 29: 139–162.
    1. 20Wang H, Bloom O, Zhang M, Vishnubhakat JM, Ombrellino M, Che J et al. HMG-1 as a late mediator of endotoxin lethality in mice. Science 1999; 285: 248–251.
    1. 21Xie J, Zhang X, Zhang L. Negative regulation of inflammation by SIRT1. Pharmacol Res 2013; 67: 60–67.
    1. 22Feige JN, Auwerx J. Transcriptional targets of sirtuins in the coordination of mammalian physiology. Curr Opin Cell Biol 2008; 20: 303–309.
    1. 23Zhang T, Kraus WL. SIRT1-dependent regulation of chromatin and transcription: linking NAD(+) metabolism and signaling to the control of cellular functions. Biochim Biophys Acta 2010; 1804: 1666–1675.
    1. 24Yeung F, Hoberg JE, Ramsey CS, Keller MD, Jones DR, Frye RA et al. Modulation of NF-kappaB-dependent transcription and cell survival by the SIRT1 deacetylase. EMBO J 2004; 23: 2369–2380.
    1. 25Zhang R, Chen HZ, Liu JJ, Jia YY, Zhang ZQ, Yang RF et al. SIRT1 suppresses activator protein-1 transcriptional activity and cyclooxygenase-2 expression in macrophages. J Biol Chem 2010; 285: 7097–7110.
    1. 26Brunet A, Sweeney LB, Sturgill JF, Chua KF, Greer PL, Lin Y et al. Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science 2004; 303: 2011–2015.
    1. 27Rajendrasozhan S, Yang SR, Kinnula VL, Rahman I. SIRT1, an antiinflammatory and antiaging protein, is decreased in lungs of patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2008; 177: 861–870.
    1. 28Shen Z, Ajmo JM, Rogers CQ, Liang X, Le L, Murr MM et al. Role of SIRT1 in regulation of LPS- or two ethanol metabolites-induced TNF-alpha production in cultured macrophage cell lines. Am J Physiol Gastrointest Liver Physiol 2009; 296: G1047–1053.
    1. 29Wang W, Lin Q, Lin R, Zhang J, Ren F, Zhang J et al. PPARα agonist fenofibrate attenuates TNF-α-induced CD40 expression in 3T3-L1 adipocytes via the SIRT1-dependent signaling pathway. Exp Cell Res 2013; 319: 1523–1533.
    1. 30Okazaki M, Iwasaki Y, Nishiyama M, Taguchi T, Tsugita M, Nakayama S et al. PPARbeta/delta regulates the human SIRT1 gene transcription via Sp1. Endocr J 2010; 57: 403–413.
    1. 31Shen Z, Liang X, Rogers CQ, Rideout D, You M. Involvement of adiponectin-SIRT1-AMPK signaling in the protective action of rosiglitazone against alcoholic fatty liver in mice. Am J Physiol Gastrointest Liver Physiol 2010; 298: G364–374.
    1. 32Haraguchi T, Takasaki K, Naito T, Hayakawa K, Katsurabayashi S, Mishima K et al. Cerebroprotective action of telmisartan by inhibition of macrophages/microglia expressing HMGB1 via a peroxisome proliferator-activated receptor gamma-dependent mechanism. Neurosci Lett 2009; 464: 151–155.
    1. 33Gao M, Hu Z, Zheng Y, Zeng Y, Shen X, Zhong D et al. Peroxisome proliferator-activated receptor γ agonist troglitazone inhibits high mobility group box 1 expression in endothelial cells via suppressing transcriptional activity of nuclear factor κB and activator protein 1. Shock 2011; 36: 228–234.
    1. 34Yang SR, Wright J, Bauter M, Seweryniak K, Kode A, Rahman I. Sirtuin regulates cigarette smoke-induced proinflammatory mediator release via RelA/p65 NF-kappaB in macrophages in vitro and in rat lungs in vivo: implications for chronic inflammation and aging. Am J Physiol Lung Cell Mol Physiol 2007; 292: L567–576.
    1. 35Kong S, Kim SJ, Sandal B, Lee SM, Gao B, Zhang DD et al. The type III histone deacetylase Sirt1 protein suppresses p300-mediated histone H3 lysine 56 acetylation at Bclaf1 promoter to inhibit T cell activation. J Biol Chem 2011; 286: 16967–16975.
    1. 36Zou T, Yang Y, Xia F, Huang A, Gao X, Fang D et al. Resveratrol Inhibits CD4+ T cell activation by enhancing the expression and activity of Sirt1. PLoS One 2013; 8: e75139.
    1. 37Cohen HY, Miller C, Bitterman KJ, Wall NR, Hekking B, Kessler B et al. Calorie restriction promotes mammalian cell survival by inducing the SIRT1 deacetylase. Science 2004; 305: 390–392.
    1. 38Chen D, Steele AD, Lindquist S, Guarente L Increase in activity during calorie restriction requires Sirt1. Science 2005; 310: 1641.
    1. 39Wang RH, Zheng Y, Kim HS, Xu X, Cao L, Luhasen T et al. Interplay among BRCA1, SIRT1, and Survivin during BRCA1-associated tumorigenesis. Mol Cell 2008; 32: 11–20.
    1. 40Chen WY, Wang DH, Yen RC, Luo J, Gu W, Baylin SB. Tumor suppressor HIC1 directly regulates SIRT1 to modulate p53-dependent DNA-damage responses. Cell 2005; 123: 437–448.
    1. 41Wang C, Chen L, Hou X, Li Z, Kabra N, Ma Y et al. Interactions between E2F1 and SirT1 regulate apoptotic response to DNA damage. Nat Cell Biol 2006; 8: 1025–1031.
    1. 42Iwahara N, Hisahara S, Hayashi T, Horio Y. Transcriptional activation of NAD+-dependent protein deacetylase SIRT1 by nuclear receptor TLX. Biochem Biophys Res Commun 2009; 386: 671–675.
    1. 43Kim MY, Kang ES, Ham SA, Hwang JS, Yoo TS, Lee H et al. The PPARδ-mediated inhibition of angiotensin II-induced premature senescence in human endothelial cells is SIRT1-dependent. Biochem Pharmacol 2012; 84: 1627–1634.
    1. 44Han L, Zhou R, Niu J, McNutt MA, Wang P, Tong T. SIRT1 is regulated by a PPAR{γ}-SIRT1 negative feedback loop associated with senescence. Nucleic Acids Res 2010; 38: 7458–7471.
    1. 45Semino C, Angelini G, Poggi A, Rubartelli A. NK/iDC interaction results in IL-18 secretion by DCs at the synaptic cleft followed by NK cell activation and release of the DC maturation factor HMGB1. Blood 2005; 106: 609–616.

Source: PubMed

3
購読する